Next Article in Journal
Efficacy of Biorational Products for Managing Diseases of Tomato in Greenhouse Production
Next Article in Special Issue
Comparative Phytochemical Analysis of Aronia melanocarpa L. Fruit Juices on Bulgarian Market
Previous Article in Journal
Optimal Deployment of WSN Nodes for Crop Monitoring Based on Geostatistical Interpolations
Previous Article in Special Issue
Therapeutic Potential of Ranunculus Species (Ranunculaceae): A Literature Review on Traditional Medicinal Herbs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Computation Screening of Multi-Target Antidiabetic Properties of Phytochemicals in Common Edible Mediterranean Plants

by
Vlasios Goulas
1,*,
Antonio J. Banegas-Luna
2,
Athena Constantinou
1,
Horacio Pérez-Sánchez
2 and
Alexandra Barbouti
3
1
Department of Agricultural Sciences, Biotechnology and Food Science, Cyprus University of Technology, Lemesos 3603, Cyprus
2
Structural Bioinformatics and High Performance Computing (BIO-HPC) Research Group, UCAM Universidad Católica de Murcia, 30107 Guadalupe, Spain
3
Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
*
Author to whom correspondence should be addressed.
Submission received: 30 May 2022 / Revised: 9 June 2022 / Accepted: 16 June 2022 / Published: 21 June 2022

Abstract

:
Diabetes mellitus is a metabolic disease and one of the leading causes of deaths worldwide. Numerous studies support that the Mediterranean diet has preventive and treatment effects on diabetes. These effects have been attributed to the special bioactive composition of Mediterranean foods. The objective of this work was to decipher the antidiabetic activity of Mediterranean edible plant materials using the DIA-DB inverse virtual screening web server. A literature review on the antidiabetic potential of Mediterranean plants was performed and twenty plants were selected for further examination. Subsequently, the most abundant flavonoids, phenolic acids, and terpenes in plant materials were studied to predict their antidiabetic activity. Results showed that flavonoids are the most active phytochemicals as they modulate the function of 17 protein-targets and present high structural similarity with antidiabetic drugs. Their antidiabetic effects are linked with three mechanisms of action, namely (i) regulation of insulin secretion/sensitivity, (ii) regulation of glucose metabolism, and (iii) regulation of lipid metabolism. Overall, the findings can be utilized to understand the antidiabetic activity of edible Mediterranean plants pinpointing the most active phytoconstituents.

1. Introduction

Diabetes mellitus (DM), or merely diabetes, is a complex chronic disease, which requires continues medical care, reducing the patient’s quality of life and raising the medical cost if not treated properly. According to the World Health Organization (WHO), DM has entered the top 10 leading causes of deaths worldwide, following a significant percentage increase of 70% from 2000 to 2030 [1]. From a medical point of view, it is a chronic noncommunicable disease arising from impaired insulin secretion and insulin resistance, leading to its defining feature of hyperglycemia. It can affect different organ systems in the body and, over time, causes several complications including neuropathy, nephropathy, retinopathy, cardiovascular disease, stroke, and peripheral artery disease [2]. The more common types of DM are classified into type 1 (diabetes mellitus (T1DM)), type 2, and gestational diabetes. Type 1 DM is characterized by absolute insulin deficiency associated with pancreatic β cells destruction [3], while type 2 DM, which accounts for nearly 95% of individuals, is mainly due to insulin resistance (IR) and deficiency in insulin secretion. Gestational diabetes, on the other hand, develops during pregnancy and usually disappears after giving birth. [3].
Recently, Newman and Cragg (2019) stated and classified all approved therapeutic agents for DM, from the 1st of January 1981 to the 30th of September 2019. According to their report, over 54% of registered drugs for DM are natural products or mimic natural products. It is obvious that natural products are an attractive reservoir of antidiabetic compounds [4]. In addition, numerous studies demonstrate an antidiabetic potential of natural products [5]. The antidiabetic effects of many phytochemicals including polyphenols, terpenes, alkaloids, saponins, and quinones have been well-documented [6,7]. Furthermore, clinical trials with medicinal plants and natural products have been conducted, whereas some of them have been used for the development of herbal formulations controlling DM. Regarding the mechanism of action of natural products, (i) the inhibition of α-glucosidase and α-amylase in the digestive tract, (ii) the boost of insulin secretion and pancreatic β cell proliferation, (iii) the regulation of glucose uptake and glucose transporters, (iv) the inhibition of protein tyrosine phosphatase 1B activity, and (v) the reduction in the generation of oxidative stress are the main modes of action of pure phytochemicals and crude extracts. [3].
Epidemiological studies also highlight that the adoption of a healthy dietary pattern such as the Mediterranean diet contributes to the prevention and treatment of Type 2 DM [8,9]. The beneficial effects of the Mediterranean diet are correlated with weight control and the consumption of foods rich in nutrients with various health benefits [10]. Fruit and vegetables are essential components of the Mediterranean diet and contain many bioactive phytochemicals. Taking into consideration a current study where researchers fished new antidiabetic compounds from various natural products, the search for new antidiabetic agents in Mediterranean edible plants stands as a challenge [5].
In the present study, we strived to clarify the antidiabetic potential of edible Mediterranean plant materials that contain common phytochemicals, with the employment of in silico virtual screening methodologies. Plants are complex mixtures of several primary and secondary metabolites and the classic approach including the isolation and the evaluation of the antidiabetic activity is time-consuming and complicated. Thus, the employment of the DIA-DB inverse virtual screening web server allows the rapid evaluation of several compounds for antidiabetic activity and predicts the possible mode of action of antidiabetic compounds.

2. Results and Discussion

2.1. Plant and Phytochemical Selection

At first, a list of plants that are widely distributed in Mediterranean flora and/or the Mediterranean diet was prepared. The in vitro and in vivo antidiabetic activity of these plants were checked with the implementation of an extensive literature review. Twenty plants were selected for further examination as they exert both in vitro and in vivo antidiabetic activity (Table 1). All selected plants are edible and/or can be consumed after cooking or processing. Regarding the plant taxonomy, they belong to 11 families. Five plants come from the Lamiaceae and four from the Apiaceae family. Both Amaryllidaceae and Asteraceae families includes two plant materials for further study. Other plant materials belong to Brassicaceae, Ranunculaceae, Plantaginaceae, Rutaceae, Rosaceae, Oleaceaceae, and Vitaceae families.
A literature review showed that the in vitro antidiabetic activity of Mediterranean plants is mostly correlated with their inhibitory effects on enzymes related to DM. About 85% of the selected plant material have α-glucosidase and/or α-amylase inhibitory activity. Both enzymes are considered as carbohydrate-hydrolyzing enzymes and are linked with postprandial hyperglycaemia as they regulate the absorption of glucose [11]. Studies also demonstrated the stimulation of insulin secretion using different pancreatic β-cells [12,13]. Coriander and black mustard seeds, thyme, and summer savory also enhance in vitro the glucose uptake in cell lines or rat muscle pieces [12,13,14]. Furthermore, in vitro studies indicated that Mediterranean plants promote the proliferation of pancreatic β-cells [13], inhibit dipeptidyl peptidase-4 inhibition [15], prevent the formation of advanced glycation end-products [16], and decrease the fat accumulation in Caenorhabditis elegans [14].
Several works also document the in vivo antidiabetic effects of selected Mediterranean plants (Table 1). This activity is mainly established using animal models such as healthy and streptozotocin or alloxan-induced diabetic rats or mice. There are also clinical trials that report their antidiabetic activity in patients with type 2 DM and overweight adults. The reduction in glucose in blood, the stimulation of insulin secretion, and the antihyperlipidemic properties are the main antidiabetic effects of selected Mediterranean plants according to the literature review. In addition, the decrease in glycosylated hemoglobin (HbA1c), the protective effects from tissue damage, and the improvement of liver functions have been also mentioned. It is noteworthy that all selected plants exert antidiabetic effects through multiple mechanisms. It is attributed to the synergism of two or more phytochemicals as plants are complex mixtures of bioactive phytochemicals.
In the next step, the phytochemical composition of active Mediterranean plants was elucidated. The constituents were classified into three groups, namely phenolic acids, flavonoids, and terpenes. The most abundant phytochemicals from each group were selected to further investigate their antidiabetic potential using the DIA-DB inverse virtual screening web server. Table 2 summarizes the phytochemicals and their distribution in the Mediterranean plants, which is as follows: quercetin is present in 19 plants, caffeic acid in 16, ferulic acid in 15, and luteolin in 15 plants. These are the most common phytochemicals in the Mediterranean plants in the present study. On the other hand, chrysoeriol, ellagic acid, and hesperidin are found less frequently in Mediterranean plants (in 5, 6, and 6 plants, respectively). All structures of the studied phytoconstituents are presented in Figure 1.

2.2. Estimation of Antidiabetic Activity of Phytochemicals using Virtual Screening

The phytochemicals were screened for antidiabetic activity against 17 diabetes targets using the DIA-DB inverse virtual screening web server. The docking scores of the crystallized ligands ranged from −10.4 to −1.5 kcal mol−1. A docking cutoff score of −8 kcal mol−1 was set, as it was deemed a reasonable average docking score that covered the most active compounds for each protein target (Table 3). Results revealed interesting findings for the antidiabetic potential of phytochemicals. More specifically, all terpenes presented docking scores bigger than −8 kcal mol−1; thus, the predicted antidiabetic activities of all terpenes are negligible or weak. The studied terpenes cannot be considered promising antidiabetic compounds for further investigation. In addition, the antidiabetic effects of edible plant materials cannot be correlated with the presence of these terpenes.
Ellagic, caffeic, and ferulic acids were active phenolic acids (Table 3). The antidiabetic potential of ellagic acid is linked with five protein targets, whereas the regulation, secretion, and/or sensitivity of insulin is the main mechanism of action of ellagic acid. In vitro and in vivo studies also reported the antidiabetic effect of ellagic acid through the stimulation of insulin [165,166]. Regarding hydroxycinnamic acids, caffeic and ferulic acids modulate the functions of only one protein target. Previous studies also manifest their antidiabetic effects by modulating insulin-signaling molecules [167,168]. A quantitative structure–activity relationship analysis for hydroxycinnamic acids will be interesting as the other acids such cinnamic acid, coumaric acid, and rosmarinic acid did not bInd. with protein targets.
Indisputably, results demonstrated that flavonoids are the most promising group of compounds. All studied flavonoids bInd. with protein targets related with DM. The antidiabetic potential of flavonoids is correlated with three possible mechanisms, namely the (i) regulation of insulin secretion and/or sensitivity, (ii) regulation of glucose metabolism, and (iii) regulation of lipid metabolism. DPP4, HSD11B1, AKR1B1, AMY2A, and PPARG protein targets interacted with the majority of flavonoids. Figure 2 shows hesperidin and rutin binds with 15 and 12 protein targets, respectively, whereas the other flavonoids modulate the function of 6 to 8 protein targets. It is noteworthy that both hesperidin and rutin are glycosylated flavonoids. Furthermore, a comparison between rutin and its aglycone form (quercetin) demonstrates the superiority of the glycosylated form. Jadhav and Puchchakayala (2012) also stated that rutin had a higher antidiabetic and hypoglycemic activity than quercetin in normoglycemic and streptozotocin -nicotinamide-induced diabetic rats performing blood glucose and serum lipid profile measurements [169]. Hesperidin is less distributed than rutin but is also a strong multi-target antidiabetic agent according to the computational measurements. These findings are in line with the literature; hesperidin is considered to have preventive and/or therapeutic effects against DM as it regulates glucose and lipid metabolism [170,171]. Overall, flavonoids are an interesting pool of antidiabetic compounds and further study is needed to probe the structural characteristics of flavonoids that are mainly linked with antidiabetic activity.

2.3. Evaluation of Molecular Similarity of Predicted Active Phytochemicals and Known/Experimental Antidiabetic Drugs

In the next step, the Tanimoto similarity index was used to quantify the similarity of the studied phytochemicals with 190 known or experimental antidiabetic drugs. A Tanimoto score of 0.7 or greater indicated a robust molecular similarity. According to our results, all tested phytochemicals have structural similarities with antidiabetic drugs. Although these findings are contrary to their predicted antidiabetic activity, the in-depth interpretation of results confirmed the previous results. More specifically, the highest molecular similarity was found for flavonoids. A previous study also correlated flavonoids with the antidiabetic activity of herbs and spices [2]. A strong molecular similarity with 132 and 131 antidiabetic drugs was detected for hesperidin (69.5%) and rutin (68.9%). Both flavonoids also bInd. with the highest number of protein targets according to our results. Furthermore, chrysoeriol (57.4%), luteolin (51.1%), and apigenin (50.5%) also showed some structural similarity with antidiabetic drugs. Similarly to the predicted antidiabetic activity, the glycosylated flavonoids had more structural similarities with antidiabetic drugs than aglycone flavonoids. From a chemical point of view, the most active flavonoids belong to the group of flavanones, flavanols, and flavones. In summary, the molecular similarity test also confirmed the potent antidiabetic effects of flavonoids.

3. Materials and Methods

3.1. Literature Review

The literature review on the antidiabetic potential of Mediterranean plants was performed using the databases of Scopus [172] and Google Scholar [173]. More specifically, the search included the terms “antidiabetic activity”, “diabetes”, “hyperglycemia”, “hypoglycemic activity”, “alpha-glucosidase”, and “alpha-amylase” in combination with “scientific plant name” and “common plant name” of Mediterranean plants. The antidiabetic potency of several plants was searched according to a list, which was prepared based on their abundance in the Mediterranean flora and/or diet.
Subsequently, data for the phytochemical composition of plants were collected using the same databases. The terms “scientific plant name”, “common plant name”, “phytochemicals”, “bioactive compounds”, “Liquid chromatography”, “Nuclear Magnetic Resonance (NMR)”, and “Liquid Chromatography-Mass Spectroscopy (LC-MS)” were used to build the compound library.

3.2. Determination of Anti-Diabetic Activity Using DIA-DB Inverse Virtual Screening Web Server

At first, a simplified molecular-input line-entry system (SMILES) notation was created for each compound, which was obtained from PubChem. [174]. All SMILES notations are demonstrated in Table 3. The SMILES notation of each compound was subsequently submitted to the DIA-DB web server that employs an inverse virtual screening of compounds with Autodock Vina against a given set of 18 protein targets associated with diabetes. The 18 protein targets can be classified into the three following groups: (i) regulation of insulin secretion and/or sensitivity (DPP4, FFAR1, HSD11B1, INSR, PTPN9, RBP4), (ii) regulation of glucose metabolism (AKR1B1, AMY2A, FBP1, GCK, MGAM, PDK2, PYGL), and (iii) regulation of lipid metabolism (NR5A2, PPARA, PPARD, PPARG, RXRA) [2,175]. More specifically, the targets were aldose reductase (AKR1B1), AMY2A, FBP1, free fatty acid receptor 1 (FFAR1), glucokinase (GCK), 11B-hydroxysteroid dehydrogenase type 1 (HSD11B1), insulin receptor (INSR), MGAM, NR5A2, pyruvate dehydrogenase kinase isoform 2 (PDK2), PPARA, PPARD, PPARG, PTPN9, liver glycogen phosphorylase (PYGL), RBP4, and retinoid X receptor alpha (RXRA). A cut-off docking score of −8 kcal mol−1 was set to distinguish between potential active and inactive compounds.

3.3. Evaluation of Molecular Similarity of Predicted Active Phytochemicals and Known/Experimental Antidiabetic Drugs

The similarities studies with known/experimental antidiabetic drugs were performed according to a previous work [176]. The molecular similarity was performed using the metric of the Tanimoto similarity on the calculated ECFP4 molecular fingerprints of the compounds. The molecular similarity network was generated with Cytoscape and the ChemViz2 Application version 1.1.0.

4. Conclusions

The present study was undertaken to shed light on the antidiabetic properties of edible Mediterranean plants. Our results showed that the most active compounds within examined Mediterranean plants are flavonoids that are widely distributed in herbs, vegetables, medicinal plants, and fruits. Our findings also show that the glycosylation of flavonoids potentially improves its antidiabetic activity. Both the evaluation of antidiabetic activity and molecular similarity of antidiabetic dugs highlighted the antidiabetic potential of hesperidin and rutin. Finally, the present study showed that the employment of the DIA-DB inverse virtual screening web server allows us to explain the antidiabetic effects of natural products and to pinpoint the most active compounds.

Author Contributions

V.G. conceived the project and designed the experiment. A.C. performed the literature review. A.J.B.-L. and H.P.-S. carried out in silico experiments. V.G., A.J.B.-L., H.P.-S., and A.B. interpreted the results and prepared the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Cyprus University Of Technology, grant number EX200146. This work was also funded by grants from the Fundación Séneca de la Región de Murcia under Project 20988/PI/18. This research was partially supported by the supercomputing infrastructure of Poznan Supercomputing Center, the e-infrastructure program of the Research Council of Norway via the supercomputer center of UiT−the Arctic University of Norway, and by the supercomputing infrastructure of the NLHPC (ECM-02), Powered@NLHPC. Finally, publication fees were covered by the Open Access Fund of Cyprus University of Technology.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. Estimates for the year 2000 and projections for 2030. World Health 2004, 27, 1047–1053. [Google Scholar]
  2. Pereira, A.S.P.; Banegas-Luna, A.J.; Peña-García, J.; Pérez-Sánchez, H.; Apostolides, Z. Evaluation of the anti-diabetic activity of some common herbs and spices: Providing new insights with inverse virtual screening. Molecules 2019, 24, 4030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Li, W.; Yuan, G.; Pan, Y.; Wang, C.; Chen, H. Network pharmacology studies on the bioactive compounds and action mechanisms of natural products for the treatment of diabetes mellitus: A review. Front. Pharmacol. 2017, 8, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef]
  5. Yeung, A.W.K.; Tzvetkov, N.T.; Durazzo, A.; Lucarini, M.; Souto, E.B.; Santini, A.; Gan, R.Y.; Jozwik, A.; Grzybek, W.; Horbańczuk, J.O.; et al. Natural products in diabetes research: Quantitative literature analysis. Nat. Prod. Res. 2021, 35, 5813–5827. [Google Scholar] [CrossRef]
  6. Xu, L.; Li, Y.; Dai, Y.; Peng, J. Natural products for the treatment of type 2 diabetes mellitus: Pharmacology and mechanisms. Pharmacol. Res. 2018, 130, 451–465. [Google Scholar] [CrossRef]
  7. Prabhakar, P.K.; Doble, M. Mechanism of action of natural products used in the treatment of diabetes mellitus. Chin. J. Integr. Med. 2011, 17, 563–574. [Google Scholar] [CrossRef]
  8. Georgoulis, M.; Kontogianni, M.D.; Yiannakouris, N. Mediterranean diet and diabetes: Prevention and treatment. Nutrients 2014, 6, 1406–1423. [Google Scholar] [CrossRef] [Green Version]
  9. Martín-Peláez, S.; Fito, M.; Castaner, O. Mediterranean diet effects on type 2 diabetes prevention, disease progression, and related mechanisms. A review. Nutrients 2020, 12, 2236. [Google Scholar] [CrossRef]
  10. Schröder, H. Protective mechanisms of the Mediterranean diet in obesity and type 2 diabetes. J. Nutr. Biochem. 2007, 18, 149–160. [Google Scholar] [CrossRef]
  11. Wang, H.; Du, Y.J.; Song, H.C. α-Glucosidase and α-amylase inhibitory activities of guava leaves. Food Chem. 2010, 123, 6–13. [Google Scholar] [CrossRef]
  12. Gray, A.M.; Flatt, P.R. Insulin-releasing and insulin-like activity of the traditional anti-diabetic plant Coriandrum sativum (coriander). Br. J. Nutr. 1999, 81, 203–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Benhaddou-Andaloussi, A.; Martineau, L.C.; Spoor, D.; Vuong, T.; Leduc, C.; Joly, E.; Burt, A.; Meddah, B.; Settaf, A.; Arnason, J.T.; et al. Antidiabetic activity of Nigella sativa seed extract in cultured pancreatic β-cells, skeletal muscle cells, and adipocytes. Pharm. Biol. 2008, 46, 96–104. [Google Scholar] [CrossRef]
  14. El-Houri, R.B.; Kotowska, D.; Olsen, L.C.B.; Bhattacharya, S.; Christensen, L.P.; Grevsen, K.; Oksbjerg, N.; Færgeman, N.; Kristiansen, K.; Christensen, K.B. Screening for bioactive metabolites in plant extracts modulating glucose uptake and fat accumulation. Evid.-Based Complement. Altern. Med. 2014, 2014, 156398. [Google Scholar] [CrossRef] [Green Version]
  15. Kalhotra, P.; Chittepu, V.C.S.R.; Osorio-Revilla, G.; Gallardo-Velazquez, T. Phytochemicals in garlic extract inhibit therapeutic enzyme DPP-4 and induce skeletal muscle cell proliferation: A possible mechanism of action to benefit the treatment of diabetes mellitus. Biomolecules 2020, 10, 305. [Google Scholar] [CrossRef] [Green Version]
  16. Perez Gutierrez, R.M. Inhibition of advanced glycation end-product formation by Origanum majorana L. in vitro and in streptozotocin-induced diabetic rats. Evid.-Based Complement. Altern. Med. 2012, 2012, 598638. [Google Scholar] [CrossRef] [Green Version]
  17. Heshmati, J.; Namazi, N. Effects of black seed (Nigella sativa) on metabolic parameters in diabetes mellitus: A systematic review. Complement. Ther. Med. 2015, 23, 275–282. [Google Scholar] [CrossRef]
  18. Benhaddou-Andaloussi, A.; Martineau, L.C.; Vallerand, D.; Haddad, Y.; Afshar, A.; Settaf, A.; Haddad, P.S. Multiple molecular targets underlie the antidiabetic effect of Nigella sativa seed extract in skeletal muscle, adipocyte and liver cells. Diabetes Obes. Metab. 2010, 12, 148–157. [Google Scholar] [CrossRef]
  19. Kaleem, M.; Kirmani, D.; Asif, M.; Ahmed, Q.; Bano, B. Biochemical effects of Nigella sativa L seeds in diabetic rats. Indian J. Exp. Biol. 2006, 44, 745–748. [Google Scholar]
  20. Agrawal, S.; Yallatikar, T.; Gurjar, P. Brassica Nigra: Ethopharmacological review of a routinely Used condiment. Curr. Drug Discov. Technol. 2018, 16, 40–47. [Google Scholar] [CrossRef]
  21. El-Manawaty, M.A.; Gohar, L. In vitro alpha-glucosidase inhibitory activity of egyptian plant extracts as an indication for their antidiabetic activity. Asian J. Pharm. Clin. Res 2018, 11, 360–367. [Google Scholar] [CrossRef]
  22. Anand, P.; Murali, K.Y.; Tandon, V.; Chandra, R.; Murthy, P.S. Preliminary studies on antihyperglycemic effect of aqueous extract of Brassica nigra (L.) Koch in streptozotocin induced diabetic rats. Indian J. Exp. Biol. 2007, 45, 696–701. [Google Scholar] [PubMed]
  23. Hawash, M.; Jaradat, N.; Elaraj, J.; Hamdan, A.; Lebdeh, S.A.; Halawa, T. Evaluation of the hypoglycemic effect of seven wild folkloric edible plants from Palestine (Antidiabetic effect of seven plants from Palestine). J. Complement. Integr. Med. 2020, 17, 1–10. [Google Scholar] [CrossRef]
  24. Thi Viet Huong, D.; Minh Giang, P.; Hoang Yen, N.; Nguyen, S.T. Plantago major L. extracts reduce blood glucose in streptozotocin-induced diabetic mice. J. Chem. 2021, 2021, 6688731. [Google Scholar] [CrossRef]
  25. Angelica Abud, M.; Nardello, A.L.; Torti, J.F. Hypoglycemic effect due to insulin stimulation with Plantago major in wistar rats. Med. Aromat. Plants 2017, 6, 292. [Google Scholar] [CrossRef] [PubMed]
  26. Lv, J.; Cao, L.; Li, M.; Zhang, R.; Bai, F.; Wei, P. Effect of hydroalcohol extract of lemon (Citrus limon) peel on a rat model of type 2 diabetes. Trop J. Pharm. Res. 2018, 17, 1367–1372. [Google Scholar] [CrossRef] [Green Version]
  27. Sathiyabama, R.G.; Rajiv Gandhi, G.; Denadai, M.; Sridharan, G.; Jothi, G.; Sasikumar, P.; de Souza Siqueira Quintans, J.; Narain, N.; Cuevas, L.E.; Coutinho, H.D.M.; et al. Evidence of insulin-dependent signalling mechanisms produced by Citrus sinensis (L.) Osbeck fruit peel in an insulin resistant diabetic animal model. Food Chem. Toxicol. 2018, 116, 86–99. [Google Scholar] [CrossRef] [Green Version]
  28. Benayad, O.; Bouhrim, M.; Tiji, S.; Kharchoufa, L.; Addi, M.; Drouet, S.; Hano, C.; Lorenzo, J.M.; Bendaha, H.; Bnouham, M.; et al. Phytochemical profile, α-glucosidase, and α-amylase inhibition potential and toxicity evaluation of extracts from Citrus aurantium (L) peel, a valuable by-product from Northeastern Morocco. Biomolecules 2021, 11, 1555. [Google Scholar] [CrossRef]
  29. Obih, P.; Obih, J.-C.; Arome, O. Is alpha-glucosidase inhibition a nechanism of the antidiabetic action of garlic (Allium sativum)? J. Biosci. Med. 2019, 7, 42–49. [Google Scholar] [CrossRef] [Green Version]
  30. Gandhi, G.R.; Vasconcelos, A.B.S.; Wu, D.T.; Li, H.B.; Antony, P.J.; Li, H.; Geng, F.; Gurgel, R.Q.; Narain, N.; Gan, R.Y. Citrus flavonoids as promising phytochemicals targeting diabetes and related complications: A systematic review of in vitro and in vivo studies. Nutrients 2020, 12, 2907. [Google Scholar] [CrossRef]
  31. Andallu, B.; Ramya, V. Anti-hyperglycemic, cholesterol-lowering and HDL-raising effects of cumin (Cuminum cyminum) seeds in type 2 diabetes. J. Nat. Remedies 2007, 7, 142–149. [Google Scholar]
  32. Srivsatava, R.; Srivastava, S.P.; Jaiswal, N.; Mishra, A.; Maurya, R.; Srivastava, A.K. Antidiabetic and antidyslipidemic activities of Cuminum cyminum L. in validated animal models. Med. Chem. Res. 2011, 20, 1656–1666. [Google Scholar] [CrossRef]
  33. Mnif, S.; Aifa, S. Cumin (Cuminum cyminum L.) from traditional uses to potential biomedical applications. Chem. Biodivers 2015, 12, 733–742. [Google Scholar] [CrossRef]
  34. Siow, H.L.; Gan, C.Y. Optimization study in extracting anti-oxidative and a-amylase inhibitor peptides from cumin seeds (Cuminum Cyminum). J. Food Biochem. 2017, 41, e12280. [Google Scholar] [CrossRef]
  35. Saleem, F.; Sarkar, D.; Ankolekar, C.; Shetty, K. Phenolic bioactives and associated antioxidant and anti-hyperglycemic functions of select species of Apiaceae family targeting for type 2 diabetes relevant nutraceuticals. Ind. Crops Prod. 2017, 107, 518–525. [Google Scholar] [CrossRef]
  36. Mishra, N. Haematological and hypoglycemic potential Anethum graveolens seeds extract in normal and diabetic Swiss albino mice. Vet. World 2013, 6, 502–507. [Google Scholar] [CrossRef]
  37. Haidari, F.; Zakerkish, M.; Borazjani, F.; Ahmadi Angali, K.; Amoochi Foroushani, G. The effects of Anethum graveolens (dill) powder supplementation on clinical and metabolic status in patients with type 2 diabetes. Trials 2020, 21, 483. [Google Scholar] [CrossRef]
  38. Eidi, A.; Eidi, M.; Esmaeili, E. Antidiabetic effect of garlic (Allium sativum L.) in normal and streptozotocin-induced diabetic rats. Phytomedicine 2006, 13, 624–629. [Google Scholar] [CrossRef]
  39. Shabani, E.; Sayemiri, K.; Mohammadpour, M. The effect of garlic on lipid profile and glucose parameters in diabetic patients: A systematic review and meta-analysis. Prim. Care Diabetes 2019, 13, 28–42. [Google Scholar] [CrossRef]
  40. Wang, J.; Zhang, X.; Lan, H.; Wang, W. Effect of garlic supplement in the management of type 2 diabetes mellitus (T2DM): A meta-analysis of randomized controlled trials. Food Nutr. Res. 2017, 61, 1377571. [Google Scholar] [CrossRef] [Green Version]
  41. Pinent, M.; Blay, M.; Bladé, M.C.; Salvadó, M.J.; Arola, L.; Ardévol, A. Grape seed-derived procyanidins have an antihyperglycemic effect in streptozotocin-induced diabetic rats and insulinomimetic activity in insulin-sensitive cell lines. Endocrinology 2004, 145, 4985–4990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Asbaghi, O.; Nazarian, B.; Reiner, Ž.; Amirani, E.; Kolahdooz, F.; Chamani, M.; Asemi, Z. The effects of grape seed extract on glycemic control, serum lipoproteins, inflammation, and body weight: A systematic review and meta-analysis of randomized controlled trials. Phyther. Res. 2020, 34, 239–253. [Google Scholar] [CrossRef] [PubMed]
  43. Bouyahya, A.; Chamkhi, I.; Benali, T.; Guaouguaou, F.E.; Balahbib, A.; El Omari, N.; Taha, D.; Belmehdi, O.; Ghokhan, Z.; El Menyiy, N. Traditional use, phytochemistry, toxicology, and pharmacology of Origanum majorana L. J. Ethnopharmacol. 2021, 265, 113318. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, Y.; Wen, M.; Zhou, P.; Tian, M.; Zhou, J.; Zhang, L. Analysis of chemical composition in Chinese olive leaf tea by UHPLC-DAD-Q-TOF-MS/MS and GC–MS and its lipid-lowering effects on the obese mice induced by high-fat diet. Food Res. Int. 2020, 128, 108785. [Google Scholar] [CrossRef]
  45. Al-Hayaly, L.; Al-Sultan, A.; Sultan, S.M.S. Effect of olive leaves extract on alloxan induced diabetes in male albino mice. In Proceedings of the 1st International Multi-Disciplinary Conference Theme: Sustainable Development and Smart Planning (IMDC-SDSP 2020), Cyberspace, 28–30 June 2020. [Google Scholar] [CrossRef]
  46. Abd El-Moneim, M.R.A.; El-Beltagi, H.S.; Fayed, S.A.; El-Ansary, A.E. Enhancing effect of olive leaves extract on lipid profile and enzymes activity in streptozotocin induced diabetic rats. Fresenius Environ. Bull. 2018, 27, 1875–1883. [Google Scholar]
  47. de Bock, M.; Derraik, J.G.B.; Brennan, C.M.; Biggs, J.B.; Morgan, P.E.; Hodgkinson, S.C.; Hofman, P.L.; Cutfield, W.S. Olive (Olea europaea L.) leaf polyphenols improve insulin sensitivity in middle-aged overweight men: A randomized, placebo-controlled, crossover trial. PLoS ONE 2013, 8, e57622. [Google Scholar] [CrossRef]
  48. Abunab, H.; Dator, W.L.; Hawamdeh, S. Effect of olive leaf extract on glucose levels in diabetes-induced rats: A systematic review and meta-analysis. J. Diabetes 2017, 9, 947–957. [Google Scholar] [CrossRef]
  49. Abouzed, T.K.; del Mar Contreras, M.; Sadek, K.M.; Shukry, M.; Abdelhady, D.H.; Gouda, W.M.; Abdo, W.; Nasr, N.E.; Mekky, R.H.; Segura-Carretero, A.; et al. Red onion scales ameliorated streptozotocin-induced diabetes and diabetic nephropathy in Wistar rats in relation to their metabolite fingerprint. Diabetes Res. Clin. Pract. 2018, 140, 253–264. [Google Scholar] [CrossRef]
  50. Kang, M.J.; Kim, J.H.; Choi, H.N.; Kim, M.J.; Han, J.H.; Lee, J.H.; Kim, J.I. Hypoglycemic effects of Welsh onion in an animal model of diabetes mellitus. Nutr. Res. Pract. 2010, 4, 486–491. [Google Scholar] [CrossRef] [Green Version]
  51. Lee, S.K.; Hwang, J.Y.; Kang, M.J.; Kim, Y.M.; Jung, S.H.; Lee, J.H.; Kim, J.I. Hypoglycemic effect of onion skin extract in animal models of diabetes mellitus. Food Sci. Biotechnol. 2008, 17, 130–134. [Google Scholar]
  52. Islam, M.S.; Choi, H.; Loots, D.T. Effects of dietary onion (Allium cepa L.) in a high-fat diet streptozotocin-induced diabetes rodent model. Ann. Nutr. Metab. 2008, 53, 6–12. [Google Scholar] [CrossRef] [PubMed]
  53. Yanardaǧ, R.; Bolkent, Ş.; Tabakoǧlu-Oǧuz, A.; Özsoy-Saçan, Ö. Effects of Petroselinum crispum extract on pancreatic B cells and blood glucose of streptozotocin-induced diabetic rats. Biol. Pharm. Bull. 2003, 26, 1206–1210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Abou Khalil, N.S.; Abou-Elhamd, A.S.; Wasfy, S.I.A.; El Mileegy, I.M.H.; Hamed, M.Y.; Ageely, H.M. Antidiabetic and antioxidant impacts of desert date (Balanites aegyptiaca) and Parsley (Petroselinum sativum) aqueous extracts: Lessons from experimental rats. J. Diabetes Res. 2016, 2016, 8408326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Cazzola, R.; Camerotto, C.; Cestaro, B. Anti-oxidant, anti-glycant, and inhibitory activity against α-amylase and α-glucosidase of selected spices and culinary herbs. Int. J. Food Sci. Nutr. 2011, 62, 175–184. [Google Scholar] [CrossRef]
  56. Agatonovic-Kustrin, S.; Kustrin, E.; Gegechkori, V.; Morton, D.W. Bioassay-guided identification of α-amylase inhibitors in herbal extracts. J. Chromatogr. A 2020, 1620, 460970. [Google Scholar] [CrossRef]
  57. Eidi, A.; Eidi, M. Antidiabetic effects of sage (Salvia officinalis L.) leaves in normal and streptozotocin-induced diabetic rats. Diabetes Metab. Syndr Clin. Res. Rev. 2009, 3, 40–44. [Google Scholar] [CrossRef]
  58. Mahdi, S.; Azzi, R.; Lahfa, F.B. Evaluation of in vitro α-amylase and α-glucosidase inhibitory potential and hemolytic effect of phenolic enriched fractions of the aerial part of Salvia officinalis L. Diabetes Metab. Syndr Clin. Res. Rev. 2020, 14, 689–694. [Google Scholar] [CrossRef]
  59. Behradmanesh, S.; Derees, F.; Kopaei, M.R. Effect of Salvia officinalis on diabetic patients. J. Ren. Inj. Prev. 2013, 2, 51–54. [Google Scholar] [CrossRef]
  60. Chen, L.; Lin, X.; Fan, X.; Qian, Y.; Lv, Q.; Teng, H. Sonchus oleraceus Linn extract enhanced glucose homeostasis through the AMPK/Akt/ GSK-3β signaling pathway in diabetic liver and HepG2 cell culture. Food Chem. Toxicol. 2020, 136, 111072. [Google Scholar] [CrossRef] [PubMed]
  61. Chen, L.; Fan, X.; Lin, X.; Qian, L.; Zengin, G.; Delmas, D.; Paoli, P.; Teng, H.; Xiao, J. Phenolic extract from Sonchus oleraceus L. protects diabetes-related liver injury in rats through TLR4/NF-κB signaling pathway. eFood 2019, 1, 77. [Google Scholar] [CrossRef] [Green Version]
  62. Kim, H.Y.; Lim, S.H.; Park, Y.H.; Ham, H.J.; Lee, K.J.; Park, D.S.; Kim, K.H.; Kim, S. Screening of α-amylase, α-glucosidase and lipase inhibitory activity with Gangwon-do wild plants extracts. J. Korean Soc Food Sci. Nutr. 2011, 40, 308–315. [Google Scholar] [CrossRef]
  63. Teugwa, C.M.; Mejiato, P.C.; Zofou, D.; Tchinda, B.T.; Boyom, F.F. Antioxidant and antidiabetic profiles of two African medicinal plants: Picralima nitida (Apocynaceae) and Sonchus oleraceus (Asteraceae). BMC Complement. Altern Med. 2013, 13, 175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. El-Hawary, S.S.; Mohammed, R.; El-Din, M.E.; Hassan, H.M.; Ali, Z.Y.; Rateb, M.E.; Bellah El Naggar, E.M.; Othman, E.M.; Abdelmohsen, U.R. Comparative phytochemical analysis of five Egyptian strawberry cultivars (Fragaria × ananassa Duch.) and antidiabetic potential of Festival and Red Merlin cultivars. RSC Adv. 2021, 11, 16755–16767. [Google Scholar] [CrossRef] [PubMed]
  65. Mandave, P.; Khadke, S.; Karandikar, M.; Pandit, V.; Ranjekar, P.; Kuvalekar, A.; Mantri, N. Antidiabetic, lipid normalizing, and nephroprotective actions of the strawberry: A potent supplementary fruit. Int. J. Mol. Sci. 2017, 18, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Da Silva Pinto, M.; de Carvalho, J.E.; Lajolo, F.M.; Genovese, M.I.; Shetty, K. Evaluation of antiproliferative, anti-type 2 diabetes, and antihypertension potentials of ellagitannins from strawberries (Fragaria × ananassa Duch.) Using In Vitro Models. J. Med. Food 2010, 13, 1027–1035. [Google Scholar] [CrossRef] [PubMed]
  67. Takács, I.; Szekeres, A.; Takács, Á.; Rakk, D.; Mézes, M.; Polyák, Á.; Lakatos, L.; Gyémánt, G.; Csupor, D.; Kovács, K.J.; et al. Wild strawberry, blackberry, and blueberry leaf extracts alleviate starch-induced hyperglycemia in prediabetic and diabetic mice. Planta Med. 2020, 86, 790–799. [Google Scholar] [CrossRef]
  68. Kemertelidze, É.P.; Sagareishvili, T.G.; Syrov, V.N.; Khushbaktova, Z.A. Chemical composition and pharmacological activity of garden savory (Satureja hortensis L.) occurring in Georgia. Pharm. Chem. J. 2004, 38, 319–322. [Google Scholar] [CrossRef]
  69. Saravanan, S.; Pari, L. Protective effect of thymol on high fat diet induced diabetic nephropathy in C57BL/6J mice. Chem. Biol. Interact. 2016, 245, 1–11. [Google Scholar] [CrossRef]
  70. Deligiannidou, G.-E.; Philippou, E.; Vidakovic, M.; Berghe, W.V.; Heraclides, A.; Grdovic, N.; Mihailovic, M.; Kontogiorgis, C. Natural products derived from the Mediterranean diet with antidiabetic Activity: From insulin mimetic hypoglycemic to nutriepigenetic modulator compounds. Curr. Pharm. Des. 2019, 25, 1760–1782. [Google Scholar] [CrossRef]
  71. Alu’datt, M.H.; Rababah, T.; Johargy, A.; Gammoh, S.; Ereifej, K.; Alhamad, M.N.; Brewer, M.S.; Saati, A.A.; Kubow, S.; Rawshdeh, M. Extraction, optimisation and characterisation of phenolics from Thymus vulgaris L.: Phenolic content and profiles in relation to antioxidant, antidiabetic and antihypertensive properties. Int. J. Food Sci. Technol. 2016, 51, 720–730. [Google Scholar] [CrossRef]
  72. Yazdanparast, R.; Ardestani, A.; Jamshidi, S. Experimental diabetes treated with Achillea santolina: Effect on pancreatic oxidative parameters. J. Ethnopharmacol. 2007, 112, 13–18. [Google Scholar] [CrossRef] [PubMed]
  73. Hamdan, I.I.; Afifi, F.U. Capillary electrophoresis as a screening tool for alpha amylase inhibitors in plant extracts. Saudi Pharm. J. 2010, 18, 91–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Al-Snafi, A.E. Chemical constituents and pharmacological activities of milfoil (Achillea santolina). A review. Int. J. PharmTech Res. 2013, 5, 1373–1377. [Google Scholar]
  75. Đorđević, B.S.; Todorović, Z.B.; Troter, D.Z.; Stanojević, L.P.; Stojanović, G.S.; Đalović, I.G.; Mitrović, P.M.; Veljković, V.B. Extraction of phenolic compounds from black mustard (Brassica nigra L.) seed by deep eutectic solvents. J. Food Meas. Charact. 2021, 15, 1931–1938. [Google Scholar] [CrossRef]
  76. Petrović, M.; Jovanović, M.; Lević, S.; Nedović, V.; Mitić-Ćulafić, D.; Semren, T.Ž.; Veljović, S. Valorization potential of Plantago major L. solid waste remaining after industrial tincture production: Insight into the chemical composition and bioactive properties. Waste Biomass Valoriz. 2022, 13, 1639–1651. [Google Scholar] [CrossRef]
  77. Singh, B.; Singh, J.P.; Kaur, A.; Singh, N. Phenolic composition, antioxidant potential and health benefits of citrus peel. Food Res. Int. 2020, 132, 109114. [Google Scholar] [CrossRef]
  78. Demir, S.; Korukluoglu, M. A comparative study about antioxidant activity and phenolic composition of cumin (Cuminum cyminum L.) and coriander (Coriandrum sativum L.). Indian J. Tradit. Knowl. 2020, 19, 383–393. [Google Scholar]
  79. Wasli, H.; Jelali, N.; Silva, A.M.S.; Ksouri, R.; Cardoso, S.M. Variation of polyphenolic composition, antioxidants and physiological characteristics of dill (Anethum graveolens L.) as affected by bicarbonate-induced iron deficiency conditions. Ind. Crops Prod. 2018, 126, 466–476. [Google Scholar] [CrossRef]
  80. Beato, V.M.; Orgaz, F.; Mansilla, F.; Montaño, A. Changes in phenolic compounds in garlic (Allium sativum L.) owing to the cultivar and location of growth. Plant. Foods Hum. Nutr. 2011, 66, 218–223. [Google Scholar] [CrossRef]
  81. Nile, S.H.; Kim, S.H.; Ko, E.Y.; Park, S.W. Polyphenolic contents and antioxidant properties of different grape (V. vinifera, V. labrusca, and V. hybrid) cultivars. Biomed. Res. Int. 2013, 2013, 718065. [Google Scholar] [CrossRef] [Green Version]
  82. Talhaoui, N.; Taamalli, A.; Gómez-Caravaca, A.M.; Fernández-Gutiérrez, A.; Segura-Carretero, A. Phenolic compounds in olive leaves: Analytical determination, biotic and abiotic influence, and health benefits. Food Res. Int. 2015, 77, 92–108. [Google Scholar] [CrossRef]
  83. Wang, H.; Provan, G.J.; Helliwell, K. Determination of rosmarinic acid and caffeic acid in aromatic herbs by HPLC. Food Chem. 2004, 87, 307–311. [Google Scholar] [CrossRef]
  84. Ferreira, F.S.; de Oliveira, V.S.; Chávez, D.W.H.; Chaves, D.S.; Riger, C.J.; Sawaya, A.C.H.F.; Guizellini, G.M.; Sampaio, G.R.; da Silva Torres, E.A.F.; Saldanha, T. Bioactive compounds of parsley (Petroselinum crispum), chives (Allium schoenoprasum L) and their mixture (Brazilian cheiro-verde) as promising antioxidant and anti-cholesterol oxidation agents in a food system. Food Res. Int. 2022, 151, 110864. [Google Scholar] [CrossRef] [PubMed]
  85. Afshari, M.; Rahimmalek, M.; Miroliaei, M. Variation in polyphenolic profiles, antioxidant and antimicrobial activity of different Achillea species as natural sources of antiglycative compounds. Chem. Biodivers. 2018, 15, e1800075. [Google Scholar] [CrossRef] [PubMed]
  86. Nour, V.; Trandafir, I.; Cosmulescu, S. Bioactive compounds, antioxidant activity and nutritional quality of different culinary aromatic herbs. Not. Bot Horti Agrobot Cluj-Napoca 2017, 45, 179–184. [Google Scholar] [CrossRef] [Green Version]
  87. Roby, M.H.H.; Sarhan, M.A.; Selim, K.A.H.; Khalel, K.I. Evaluation of antioxidant activity, total phenols and phenolic compounds in thyme (Thymus vulgaris L.), sage (Salvia officinalis L.), and marjoram (Origanum majorana L.) extracts. Ind. Crops Prod. 2013, 43, 827–831. [Google Scholar] [CrossRef]
  88. Kobeasy, I.; Abdel-Fatah, M.; Abd El-Salam, S.M.; El-Ola Mohamed, Z.M. Biochemical studies on Plantago major L. and Cyamopsis tetragonoloba L. Int. J. Biodivers. Conserv. 2011, 3, 83–91. [Google Scholar]
  89. Mohanty, S.; Maurya, A.K.; Jyotshna; Saxena, A.; Shanker, K.; Pal, A.; Bawankule, D.U. Flavonoids rich fraction of Citrus limetta fruit peels reduces proinflammatory cytokine production and attenuates Malaria pathogenesis. Curr. Pharm. Biotechnol. 2015, 16, 544–552. [Google Scholar] [CrossRef]
  90. Sandhu, A.K.; Gu, L. Antioxidant capacity, phenolic content, and profiling of phenolic compounds in the seeds, skin, and pulp of Vitis rotundifolia (Muscadine Grapes) as determined by HPLC-DAD-ESI-MSn. J. Agric. Food Chem. 2010, 58, 4681–4692. [Google Scholar] [CrossRef]
  91. Saleh, A.M.; Selim, S.; Al Jaouni, S.; AbdElgawad, H. CO2 enrichment can enhance the nutritional and health benefits of parsley (Petroselinum crispum L.) and dill (Anethum graveolens L.). Food Chem. 2018, 269, 519–526. [Google Scholar] [CrossRef]
  92. Williner, M.R.; Pirovani, M.E.; Güemes, D.R. Ellagic acid content in strawberries of different cultivars and ripening stages. J. Sci. Food Agric. 2003, 83, 842–845. [Google Scholar] [CrossRef]
  93. Bourgou, S.; Ksouri, R.; Bellila, A.; Skandrani, I.; Falleh, H.; Marzouk, B. Phenolic composition and biological activities of Tunisian Nigella sativa L. shoots and roots. Comptes Rendus Biol. 2008, 331, 48–55. [Google Scholar] [CrossRef]
  94. Prakash, D.; Singh, B.N.; Upadhyay, G. Antioxidant and free radical scavenging activities of phenols from onion (Allium cepa). Food Chem. 2007, 102, 1389–1393. [Google Scholar] [CrossRef]
  95. Barbieri, J.B.; Goltz, C.; Batistão Cavalheiro, F.; Theodoro Toci, A.; Igarashi-Mafra, L.; Mafra, M.R. Deep eutectic solvents applied in the extraction and stabilization of rosemary (Rosmarinus officinalis L.) phenolic compounds. Ind. Crops Prod. 2020, 144, 112049. [Google Scholar] [CrossRef]
  96. Russell, W.R.; Scobbie, L.; Labat, A.; Duthie, G.G. Selective bio-availability of phenolic acids from Scottish strawberries. Mol. Nutr. Food Res. 2009, 53, 85–91. [Google Scholar] [CrossRef]
  97. Rebey, I.B.; Zakhama, N.; Karoui, I.J.; Marzouk, B. Polyphenol composition and antioxidant activity of cumin (Cuminum Cyminum L.) seed extract under drought. J. Food Sci. 2012, 77, C734–C739. [Google Scholar] [CrossRef]
  98. Fratianni, F.; Ombra, M.N.; Cozzolino, A.; Riccardi, R.; Spigno, P.; Tremonte, P.; Coppola, R.; Nazzaro, F. Phenolic constituents, antioxidant, antimicrobial and anti-proliferative activities of different endemic Italian varieties of garlic (Allium sativum L.). J. Funct. Foods 2016, 21, 240–248. [Google Scholar] [CrossRef]
  99. Obreque-Slier, E.; Herrera-Bustamante, B.; López-Solís, R. Ripening-associated flattening out of inter-varietal differences in some groups of phenolic compounds in the skins of six emblematic grape wine varieties. J. Food Compos. Anal. 2021, 99, 103858. [Google Scholar] [CrossRef]
  100. Yilmaz, Y.; Toledo, R.T. Major Flavonoids in grape seeds and skins: Antioxidant capacity of catechin, epicatechin, and gallic acid. J. Agric. Food Chem. 2004, 52, 255–260. [Google Scholar] [CrossRef]
  101. Mahmood, T.; Anwar, F.; Abbas, M.; Saari, N. Effect of maturity on phenolics (Phenolic acids and flavonoids) profile of strawberry cultivars and mulberry species from Pakistan. Int. J. Mol. Sci. 2012, 13, 4591–4607. [Google Scholar] [CrossRef] [Green Version]
  102. Erdogan Orhan, I.; Senol, F.S.; Ozturk, N.; Celik, S.A.; Pulur, A.; Kan, Y. Phytochemical contents and enzyme inhibitory and antioxidant properties of Anethum graveolens L. (dill) samples cultivated under organic and conventional agricultural conditions. Food Chem. Toxicol. 2013, 59, 96–103. [Google Scholar] [CrossRef] [PubMed]
  103. Tepe, B.; Sokmen, A. Production and optimisation of rosmarinic acid by Satureja hortensis L. callus cultures. Nat. Prod. Res. 2007, 21, 1133–1144. [Google Scholar] [CrossRef] [PubMed]
  104. Poureini, F.; Mohammadi, M.; Najafpour, G.D.; Nikzad, M. Comparative study on the extraction of apigenin from parsley leaves (Petroselinum crispum L.) by ultrasonic and microwave methods. Chem. Pap. 2020, 74, 3857–3871. [Google Scholar] [CrossRef]
  105. Singh, B.; Singh, J.P.; Kaur, A.; Yadav, M.P. Insights into the chemical composition and bioactivities of citrus peel essential oils. Food Res. Int. 2021, 143, 110231. [Google Scholar] [CrossRef] [PubMed]
  106. Alrekabi, D.G.; Hamad, M.N. Phytochemical investigation of Sonchus oleraceus (Family:Asteraceae) cultivated in Iraq, isolation and identification of quercetin and apigenin. J. Pharm. Sci. Res. 2018, 10, 2242–2248. [Google Scholar]
  107. Boroja, T.; Katanić, J.; Rosić, G.; Selaković, D.; Joksimović, J.; Mišić, D.; Stanković, V.; Jovičić, N.; Mihailović, V. Summer savory (Satureja hortensis L.) extract: Phytochemical profile and modulation of cisplatin-induced liver, renal and testicular toxicity. Food Chem. Toxicol. 2018, 118, 252–263. [Google Scholar] [CrossRef]
  108. Lee, Y.H.; Choo, C.; Waisundara, V.Y. Determination of the total antioxidant capacity and quantification of phenolic compounds of different solvent extracts of black mustard seeds (Brassica nigra). Int. J. Food Prop. 2015, 18, 2500–2507. [Google Scholar] [CrossRef]
  109. Indah, K.P.; Menara, B.; Mpaj, P.; Utama, J.P.; Lumpur, K. GC-MS Analysis of various extracts from leaf of Plantago major used as traditional medicine. World Appl. Sci. J. 2012, 17, 67–70. [Google Scholar]
  110. Insani, E.M.; Cavagnaro, P.F.; Salomón, V.M.; Langman, L.; Sance, M.; Pazos, A.A.; Carrari, F.O.; Filippini, O.; Vignera, L.; Galmarini, C.R. Variation for health-enhancing compounds and traits in onion (Allium cepa L.) germplasm. Food Nutr. Sci. 2016, 7, 577–591. [Google Scholar] [CrossRef] [Green Version]
  111. Kelebek, H.; Selli, S. Characterization of phenolic compounds in strawberry fruits by RP-HPLC-DAD and investigation of their antioxidant capacity. J. Liq. Chromatogr. Relat. Technol. 2011, 34, 2495–2504. [Google Scholar] [CrossRef]
  112. Soliman, M.A.; Galal, T.M.; Naim, M.A.; Khalafallah, A.A. Seasonal variation in the secondary metabolites and antimicrobial activity of Plantago major L. from Egyptian heterogenic habitats. Egypt J. Bot. 2022, 62, 255–273. [Google Scholar] [CrossRef]
  113. El-Sayed, M.A.; Al-Gendy, A.A.; Hamdan, D.I.; El-Shazly, A.M. Phytoconstituents, LC-ESI-MS profile, antioxidant and antimicrobial activities of Citrus x limon L. Burm. F. cultivar variegated pink lemon. J. Pharm. Sci. Res. 2017, 9, 375–391. [Google Scholar]
  114. Oganesyan, E.T.; Nersesyan, Z.M.; Parkhomenko, A.Y. Chemical composition of the above-ground part of Coriandrum sativum. Pharm. Chem. J. 2007, 41, 149–153. [Google Scholar] [CrossRef]
  115. Pieroni, A.; Heimler, D.; Pieters, L.; van Poel, B.; Vlietinck, A.J. In vitro anti-complementary activity of flavonoids from olive (Olea europaea L.) leaves. Pharmazie 1996, 51, 765–768. [Google Scholar]
  116. Zeng, X.; Shi, J.; Zhao, M.; Chen, Q.; Wang, L.; Jiang, H.; Luo, F.; Zhu, L.; Lu, L.; Wang, X.; et al. Regioselective glucuronidation of diosmetin and chrysoeriol by the interplay of glucuronidation and transport in UGT1A9-Overexpressing hela cells. PLoS ONE 2016, 11, e0166239. [Google Scholar] [CrossRef]
  117. Nie, J.Y.; Li, R.; Wang, Y.; Tan, J.; Tang, S.H.; Jiang, Z.T. Antioxidant activity evaluation of rosemary ethanol extract and their cellular antioxidant activity toward HeLa cells. J. Food Biochem. 2019, 43, e12851. [Google Scholar] [CrossRef]
  118. Mustafa, A.M.; Angeloni, S.; Abouelenein, D.; Acquaticci, L.; Xiao, J.; Sagratini, G.; Maggi, F.; Vittori, S.; Caprioli, G. A new HPLC-MS/MS method for the simultaneous determination of 36 polyphenols in blueberry, strawberry and their commercial products and determination of antioxidant activity. Food Chem. 2022, 367, 130743. [Google Scholar] [CrossRef]
  119. Chkhikvishvili, I.; Sanikidze, T.; Gogia, N.; McHedlishvili, T.; Enukidze, M.; Machavariani, M.; Vinokur, Y.; Rodov, V. Rosmarinic acid-rich extracts of summer savory (Satureja hortensis L.) protect jurkat T cells against oxidative stress. Oxid. Med. Cell Longev. 2013, 2013, 456253. [Google Scholar] [CrossRef] [Green Version]
  120. Chen, Y.; Wen, J.; Deng, Z.; Pan, X.; Xie, X.; Peng, C. Effective utilization of food wastes: Bioactivity of grape seed extraction and its application in food industry. J. Funct. Foods 2020, 73, 104113. [Google Scholar] [CrossRef]
  121. Stan, M.; Soran, M.L.; Varodi, C.; Lung, I. Extraction and identification of flavonoids from parsley extracts by HPLC analysis. AIP Conf. Proc. 2012, 1425, 50–52. [Google Scholar] [CrossRef]
  122. Vallverdú-Queralt, A.; Regueiro, J.; Martínez-Huélamo, M.; Rinaldi Alvarenga, J.F.; Leal, L.N.; Lamuela-Raventos, R.M. A comprehensive study on the phenolic profile of widely used culinary herbs and spices: Rosemary, thyme, oregano, cinnamon, cumin and bay. Food Chem. 2014, 154, 299–307. [Google Scholar] [CrossRef]
  123. Mašković, P.; Veličković, V.; Mitić, M.; Đurović, S.; Zeković, Z.; Radojković, M.; Cvetanović, A.; Švarc-Gajić, J.; Vujić, J. Summer savory extracts prepared by novel extraction methods resulted in enhanced biological activity. Ind. Crops Prod. 2017, 109, 875–881. [Google Scholar] [CrossRef]
  124. Wojdyło, A.; Oszmiański, J.; Czemerys, R. Antioxidant activity and phenolic compounds in 32 selected herbs. Food Chem. 2007, 105, 940–949. [Google Scholar] [CrossRef]
  125. Hasan, F.; Aziz, S.A.; Melati, M. Leaf and flavonoid production of perennial sow-thistle (Sonchus arvensis L.) at different growth stages. Int. J. Biosci. 2017, 10, 147–155. [Google Scholar] [CrossRef]
  126. Świąder, K.; Hallmann, E.; Piotrowska, A. The effect of organic practices on the bioactive compounds content in strawberry fruits. J. Res. Appl. Agric. Eng. 2016, 61, 176–179. [Google Scholar]
  127. Kulišić, T.; Kriško, A.; Dragovic-Uzelać, V.; Miloš, M.; Pifat, G. The effects of essential oils and aqueous tea infusions of oregano (Origanum vulgare L. spp. hirtum), thyme (Thymus vulgaris L.) and wild thyme (Thymus serpyllum L.) on the copper-induced oxidation of human low-density lipoproteins. Int. J. Food Sci. Nutr. 2007, 58, 87–93. [Google Scholar] [CrossRef] [PubMed]
  128. Bianchin, M.; Pereira, D.; de Florio Almeida, J.; de Moura, C.; Pinheiro, R.S.; Heldt, L.F.S.; Haminiuk, C.W.I.; Carpes, S.T. Antioxidant properties of lyophilized rosemary and sage extracts and its effect to prevent lipid oxidation in poultry pátê. Molecules 2020, 25, 5160. [Google Scholar] [CrossRef]
  129. Herrera, M.C.; Luque De Castro, M.D. Ultrasound-assisted extraction for the analysis of phenolic compounds in strawberries. Anal. Bioanal. Chem. 2004, 379, 1106–1112. [Google Scholar] [CrossRef]
  130. Mocan, A.; Babotă, M.; Pop, A.; Fizeșan, I.; Diuzheva, A.; Locatelli, M.; Carradori, S.; Campestre, C.; Menghini, L.; Sisea, C.R.; et al. Chemical constituents and biologic activities of sage species: A comparison between Salvia officinalis L., S. glutinosa L and S. transsylvanica (schur ex griseb. & schenk) schur. Antioxidants 2020, 9, 480. [Google Scholar] [CrossRef]
  131. Al-Maqtari, M.A.A.; Alghalibi, S.M.; Alhamzy, E.H. Chemical composition and antimicrobial activity of essential oil of Thymus vulgaris from yemen. Turkish J. Biochem. 2011, 36, 342–349. [Google Scholar]
  132. Lakwani, M.A.S.; Kenanoğlu, O.N.; Taştan, Y.; Bilen, S. Effects of black mustard (Brassica nigra) seed oil on growth performance, digestive enzyme activities and immune responses in rainbow trout (Oncorhynchus mykiss). Aquac. Res. 2022, 53, 300–313. [Google Scholar] [CrossRef]
  133. Lasram, S.; Zemni, H.; Hamdi, Z.; Chenenaoui, S.; Houissa, H.; Saidani Tounsi, M.; Ghorbel, A. Antifungal and antiaflatoxinogenic activities of Carum carvi L., Coriandrum sativum L. seed essential oils and their major terpene component against Aspergillus flavus. Ind. Crops Prod. 2019, 134, 11–18. [Google Scholar] [CrossRef]
  134. Ben Miri, Y.; Djenane, D. Evaluation of protective impact of algerian Cuminum cyminum L. and Coriandrum sativum L. essential oils on Aspergillus flavus growth and aflatoxin B1 production. Pakistan J. Biol. Sci. 2018, 21, 67–77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Kaur, V.; Kaur, R.; Bhardwaj, U. A review on dill essential oil and its chief compounds as natural biocide. Flavour Fragr. J. 2021, 36, 412–431. [Google Scholar] [CrossRef]
  136. Wu, Y.; Duan, S.; Zhao, L.; Gao, Z.; Luo, M.; Song, S.; Xu, W.; Zhang, C.; Ma, C.; Wang, S. Aroma characterization based on aromatic series analysis in table grapes. Sci. Rep. 2016, 6, 31116. [Google Scholar] [CrossRef] [Green Version]
  137. Konoz, E.; Abbasi, A.; Moazeni, R.S.; Parastar, H.; Jalali-Heravi, M. Chemometrics-assisted gas chromatographic-mass spectrometric analysis of volatile components of olive leaf oil. J. Iran. Chem. Soc. 2013, 10, 169–179. [Google Scholar] [CrossRef]
  138. Woo, J.-H.; Mok, M.-G.; Han, K.-W.; Lee, S.-Y.; Park, K.-W. Aroma components and Aatioxidant activities of pure rosemary essential oil goods produced in different countries. Korean J. Hortic Sci. Technol. 2010, 28, 696–700. [Google Scholar]
  139. Skubij, N.; Dzida, K. Essential oil composition of summer savory (Satureja hortensis L.) cv. Saturn depending on nitrogen nutrition and plant development phases in raw material cultivated for industrial use. Ind. Crops Prod. 2019, 135, 260–270. [Google Scholar] [CrossRef]
  140. Dubey, P.N.; Saxena, S.N.; Mishra, B.K.; Solanki, R.K.; Vishal, M.K.; Singh, B.; Sharma, L.K.; John, S.; Agarwal, D.; Yogi, A. Preponderance of cumin (Cuminum cyminum L.) essential oil constituents across cumin growing agro-ecological sub regions, India. Ind. Crops Prod. 2017, 95, 50–59. [Google Scholar] [CrossRef]
  141. Craft, J.D.; Setzer, W.N. The volatile components of parsley, Petroselinum crispum (Mill.) Fuss. Am. J. Essent. Oils Nat. Prod. 2017, 5, 27–32. [Google Scholar]
  142. Jiang, Y.; Wu, N.; Fu, Y.J.; Wang, W.; Luo, M.; Zhao, C.J.; Zu, Y.G.; Liu, X.L. Chemical composition and antimicrobial activity of the essential oil of Rosemary. Environ. Toxicol. Pharmacol. 2011, 32, 63–68. [Google Scholar] [CrossRef] [PubMed]
  143. Çalişkan, T.; Maral, H.; Pala, C.; Kafkas, E.; Kirici, S. Morphogenetic variation for essential oil content and composition of sage ( Salvia officinalis L.) In Çukurova condition introduction. Arab. J. Med. Aromat. Plants 2019, 5, 32–38. [Google Scholar]
  144. Mohtashami, S.; Rowshan, V.; Tabrizi, L.; Babalar, M.; Ghani, A. Summer savory (Satureja hortensis L.) essential oil constituent oscillation at different storage conditions. Ind. Crops Prod. 2018, 111, 226–231. [Google Scholar] [CrossRef]
  145. Hudaib, M.; Speroni, E.; Di Pietra, A.M.; Cavrini, V. GC/MS evaluation of thyme (Thymus vulgaris L.) oil composition and variations during the vegetative cycle. J. Pharm. Biomed. Anal. 2002, 29, 691–700. [Google Scholar] [CrossRef]
  146. El-Shazly, A.M.; Hafez, S.S.; Wink, M. Comparative study of the essential oils and extracts of Achillea fragrantissima (Forssk.) Sch. Bip. and Achillea santolina L. (Asteraceae) from Egypt. Pharmazie 2004, 59, 226–230. [Google Scholar]
  147. Palmieri, S.; Pellegrini, M.; Ricci, A.; Compagnone, D.; lo Sterzo, C. Chemical composition and antioxidant activity of thyme, hemp and coriander extracts: A comparison study of maceration, soxhlet, UAE and RSLDE techniques. Foods 2020, 9, 1221. [Google Scholar] [CrossRef]
  148. Patel, B.D.; Bhadada, S.V.; Ghate, M.D. Design, synthesis and anti-diabetic activity of triazolotriazine derivatives as dipeptidyl peptidase-4 (DPP-4) inhibitors. Bioorg. Chem. 2017, 72, 345–358. [Google Scholar] [CrossRef]
  149. Governa, P.; Caroleo, M.C.; Carullo, G.; Aiello, F.; Cione, E.; Manetti, F. FFAR1/GPR40: One target, different binding sites, many agonists, no drugs, but a continuous and unprofitable tug-of-war between ligand lipophilicity, activity, and toxicity. Bioorg. Med. Chem. Lett. 2021, 41, 127969. [Google Scholar] [CrossRef]
  150. Nguyen Vo, T.H.; Tran, N.; Nguyen, D.; Le, L. An in silico study on antidiabetic activity of bioactive compounds in Euphorbia thymifolia Linn. Springerplus 2016, 5, 1359. [Google Scholar] [CrossRef] [Green Version]
  151. Kong, W.J.; Zhang, H.; Song, D.Q.; Xue, R.; Zhao, W.; Wei, J.; Wang, Y.M.; Shan, N.; Zhou, Z.X.; Yang, P.; et al. Berberine reduces insulin resistance through protein kinase C-dependent up-regulation of insulin receptor expression. Metabolism 2009, 58, 109–119. [Google Scholar] [CrossRef]
  152. Sharma, C.; Kim, Y.; Ahn, D.; Chung, S.J. Protein tyrosine phosphatases (PTPs) in diabetes: Causes and therapeutic opportunities. Arch. Pharm. Res. 2021, 44, 310–321. [Google Scholar] [CrossRef] [PubMed]
  153. Liu, J.; Song, C.; Nie, C.; Sun, Y.; Wang, Y.; Xue, L.; Fan, M.; Qian, H.; Wang, L.; Li, Y. A novel regulatory mechanism of geniposide for improving glucose homeostasis mediated by circulating RBP4. Phytomedicine 2022, 95, 153862. [Google Scholar] [CrossRef]
  154. Reddy, A.B.M.; Tammali, R.; Mishra, R.; Srivastava, S.; Srivastava, S.K.; Ramana, K.V. Aldose reductase deficiency protects sugar-induced lens opacification in rats. Chem. Biol. Interact. 2011, 191, 346–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Usher, C.L.; Handsaker, R.E.; Esko, T.; Tuke, M.A.; Weedon, M.N.; Hastie, A.R.; Cao, H.; Moon, J.E.; Kashin, S.; Fuchsberger, C.; et al. Structural forms of the human amylase locus and their relationships to SNPs, haplotypes and obesity. Nat. Genet. 2015, 47, 921–925. [Google Scholar] [CrossRef] [PubMed]
  156. Petersen, M.C.; Vatner, D.F.; Shulman, G.I. Regulation of hepatic glucose metabolism in health and disease. Nat. Rev. Endocrinol. 2017, 13, 572–587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Lee, B.H.; Eskandari, R.; Jones, K.; Reddy, K.R.; Quezada-Calvillo, R.; Nichols, B.L.; Rose, D.R.; Hamaker, B.R.; Pinto, B.M. Modulation of starch digestion for slow glucose release through “Toggling” of activities of mucosal α-glucosidases. J. Biol. Chem. 2012, 287, 31929–31938. [Google Scholar] [CrossRef] [Green Version]
  158. Sugden, M.C.; Holness, M.J. Recent advances in mechanisms regulating glucose oxidation at the level of the pyruvate dehydrogenase complex by PDKs. Am. J. Physiol. Endocrinol. Metab. 2003, 284, E855–E862. [Google Scholar] [CrossRef] [Green Version]
  159. Zois, C.E.; Favaro, E.; Harris, A.L. Glycogen metabolism in cancer. Biochem. Pharmacol. 2014, 92, 3–11. [Google Scholar] [CrossRef]
  160. Fayard, E.; Auwerx, J.; Schoonjans, K. LRH-1: An orphan nuclear receptor involved in development, metabolism and steroidogenesis. Trends Cell Biol. 2004, 14, 250–260. [Google Scholar] [CrossRef]
  161. Bionaz, M.; Thering, B.J.; Loor, J.J. Fine metabolic regulation in ruminants via nutrient-gene interactions: Saturated long-chain fatty acids increase expression of genes involved in lipid metabolism and immune response partly through PPAR-α activation. Br. J. Nutr. 2012, 107, 179–191. [Google Scholar] [CrossRef]
  162. Luquet, S.; Lopez-Soriano, J.; Holst, D.; Gaudel, C.; Jehl-Pietri, C.; Fredenrich, A.; Grimaldi, P.A. Roles of peroxisome proliferator-activated receptor delta (PPARδ) in the control of fatty acid catabolism. A new target for the treatment of metabolic syndrome. Biochimie 2004, 86, 833–837. [Google Scholar] [CrossRef] [PubMed]
  163. Janani, C.; Ranjitha Kumari, B.D. PPAR gamma gene - A review. Diabetes Metab. Syndr Clin. Res. Rev. 2015, 9, 46–50. [Google Scholar] [CrossRef] [PubMed]
  164. Grzegorzewska, A.E.; Niepolski, L.; Mostowska, A.; Warchoł, W.; Jagodziński, P.P. Involvement of adropin and adropin-associated genes in metabolic abnormalities of hemodialysis patients. Life Sci. 2016, 160, 41–46. [Google Scholar] [CrossRef] [PubMed]
  165. Harakeh, S.; Almuhayawi, M.; Al Jaouni, S.; Almasaudi, S.; Hassan, S.; Al Amri, T.; Azhar, N.; Abd-Allah, E.; Ali, S.; El-Shitany, N.; et al. Antidiabetic effects of novel ellagic acid nanoformulation: Insulin-secreting and anti-apoptosis effects. Saudi J. Biol. Sci. 2020, 27, 3474–3480. [Google Scholar] [CrossRef]
  166. Fatima, N.; Hafizur, R.M.; Hameed, A.; Ahmed, S.; Nisar, M.; Kabir, N. Ellagic acid in Emblica officinalis exerts anti-diabetic activity through the action on β-cells of pancreas. Eur. J. Nutr. 2017, 56, 591–601. [Google Scholar] [CrossRef]
  167. Huang, D.A.W.; Shen, S.C.; Swi-Bea Wu, J. Effects of caffeic acid and cinnamic acid on glucose uptake in insulin-resistant mouse hepatocytes. J. Agric. Food Chem. 2009, 57, 7687–7692. [Google Scholar] [CrossRef]
  168. Narasimhan, A.; Chinnaiyan, M.; Karundevi, B. Insulin-signalling molecules in the liver of high-fat diet and fructose-induced type-2 diabetic adult male rat. Appl. Physiol. Nutr. Metab. 2015, 40, 769–781. [Google Scholar] [CrossRef]
  169. Jadhav, R.; Puchchakayala, G. Hypoglycemic and antidiabetic activity of flavonoids: Boswellic acid, ellagic acid, quercetin, rutin on streptozotocin-nicotinamide iduced type 2 diabetic rats. Int. J. Pharm. Pharm. Sci 2012, 4, 2–7. [Google Scholar]
  170. Xiong, H.; Wang, J.; Ran, Q.; Lou, G.; Peng, C.; Gan, Q.; Hu, J.; Sun, J.; Yao, R.; Huang, Q. Hesperidin: A therapeutic agent for obesity. Drug Des. Devel Ther. 2019, 13, 3855–3866. [Google Scholar] [CrossRef] [Green Version]
  171. Yoshida, H.; Tsuhako, R.; Sugita, C.; Kurokawa, M. Glucosyl hesperidin has an anti-diabetic effect in high-fat diet-induced obese mice. Biol. Pharm. Bull. 2021, 44, 422–430. [Google Scholar] [CrossRef]
  172. Available online: https://0-www-scopus-com.brum.beds.ac.uk/search/form.uri?display=basic#basic (accessed on 20 April 2022).
  173. Available online: https://0-scholar-google-com.brum.beds.ac.uk/ (accessed on 20 April 2022).
  174. Available online: https://pubchem.ncbi.nlm.nih.gov/ (accessed on 10 March 2021).
  175. Pérez-Sánchez, H.; den-Haan, H.; Peña-García, J.; Lozano-Sánchez, J.; Martínez Moreno, M.E.; Sánchez-Pérez, A.; Muñoz, A.; Ruiz-Espinosa, P.; Pereira, A.S.P.; Katsikoudi, A.; et al. DIA-DB: A database and web server for the prediction of diabetes drugs. J. Chem. Inf. Model. 2020, 60, 4124–4130. [Google Scholar] [CrossRef] [PubMed]
  176. Pereira, A.S.P.; den Haan, H.; Peña-García, J.; Moreno, M.M.; Pérez-Sánchez, H.; Apostolides, Z. Exploring african medicinal plants for potential anti-diabetic compounds with the DIA-DB inverse virtual screening web server. Molecules 2019, 24, 2002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Structures of the studied phytochemicals present in Mediterranean plants.
Figure 1. Structures of the studied phytochemicals present in Mediterranean plants.
Plants 11 01637 g001
Figure 2. Interaction of hesperidin and rutin with protein targets related with diabetes mellitus as predicted with the employment of DIA-DB inverse virtual screening web server.
Figure 2. Interaction of hesperidin and rutin with protein targets related with diabetes mellitus as predicted with the employment of DIA-DB inverse virtual screening web server.
Plants 11 01637 g002
Table 1. A comprehensive summary of in vitro and in vivo antidiabetic properties of edible Mediterranean plants.
Table 1. A comprehensive summary of in vitro and in vivo antidiabetic properties of edible Mediterranean plants.
Common NameScientific NamePlant PartIn vitro Antidiabetic EffectsIn vivo Antidiabetic EffectsReferences
Black cuminNigella sativa L.Seeds▪ Increase in insulin secretion
▪ Induction in proliferation of pancreatic β-cells
▪ Stimulation of glucose uptake
▪ Reduction in blood glucose
▪ Antihyperlipidemic effects
[13,17,18,19]
Black mustardBrassica nigra L.Aerial plants, seeds▪ α-glucosidase inhibition▪ Reduction in blood glucose
▪ Decrease in glycosylated hemoglobin (HbA1c)
▪ Antihyperlipidemic effects
▪ Insulinotropic effect
[20,21,22]
Broadleaf plantainPlantago major L.Leaves▪ α-amylase inhibition▪ Reduction in blood glucose
▪ Stimulation of insulin secretion
[23,24,25]
Citrus fruitsCitrus spp.Peel▪ α-glucosidase inhibition
▪ α-amylase inhibition
▪ Reduction in blood glucose
▪ Antihyperlipidemic effects
▪ Stimulation of insulin secretion
▪ Protective effects for tissue damage
[26,27,28,29,30]
CorianderCoriandrum sativum L.Seeds▪ Stimulation of insulin secretion
▪ Enhancement of glucose uptake
▪ Stimulation of insulin secretion
▪ Enhancement of glucose uptake
▪ Reduction in blood glucose
▪ Antihyperlipidemic effects
[12]
CuminCuminum cyminum L.Seeds▪ α-glucosidase inhibition
▪ α-amylase inhibition
▪ Reduction in blood glucose
▪ Antihyperlipidemic effects
▪ Decrease of HbA1c
[31,32,33,34]
DillAnethum graveolens L.Seeds▪ α-glucosidase inhibition
▪ α-amylase inhibition
▪ Reduction in blood glucose
▪ Antihyperlipidemic effects
▪ Control of colonic motility disorder
[35,36,37]
GarlicAllium sativum LBulb▪ dipeptidyl peptidase-4 inhibition
▪ α-glucosidase inhibition
▪ Reduction in blood glucose
▪ Antihyperlipidemic effects
▪ Decrease of insulin resistance
[15,38,39,40]
GrapesVitis vinifera L.Seeds, skinsα-glucosidase inhibition α-amylase inhibition▪ Reduction in blood glucose
▪ Antihyperlipidemic effects
▪ Decrease in HbA1c
[41,42]
MarjoramOriganum majorana L.Aerial parts▪ α-glucosidase inhibition
▪ Inhibition of advanced glycation end-product (AGE) formation
▪ Inhibition of (AGE) formation
▪ Reduction in blood glucose
▪ Antihyperlipidemic effects
[16,43]
OliveOlea europaea L.Leaves▪ α-glucosidase inhibition
▪ α-amylase inhibition
▪ Reduction in blood glucose
▪ Stimulation of insulin secretion
▪ Antihyperlipidemic effects
▪ Decrease in histopathological changes
▪ α-amylase inhibition
[44,45,46,47,48]
OnionAllium sepa L.Bulbs, skins▪ α-glucosidase inhibition ▪ Reduction in blood glucose
▪ Stimulation of insulin secretion
[49,50,51,52]
ParsleyPetroselinum sativum/crispumLeaves▪ α-glucosidase inhibition
▪ α-amylase inhibition
▪ Reduction in blood glucose
▪ Stimulation of insulin secretion
▪ Antihyperlipidemic effects
[53,54,55]
RosemaryRosmarinus officinalis L.Aerial parts▪ α-glucosidase inhibition ▪ Stimulation of insulin secretion[56]
SageSalvia officinalis L.Aerial parts▪ α-glucosidase inhibition
▪ α-amylase inhibition
▪ Reduction in blood glucose
▪ Stimulation of insulin secretion
▪ Antihyperlipidemic effects
[57,58,59]
Sow thistleSonchus oleraceus L. ▪ α-glucosidase inhibition
▪ α-amylase inhibition
▪ Reduction in blood glucose
▪ Stimulation of insulin secretion
▪ Antihyperlipidemic effects
▪ Protective effects for tissue damage
[60,61,62,63]
StrawberryFragaria spp.Leaves, fruits▪ α-glucosidase inhibition
▪ α-amylase inhibition
▪ Reduction in blood glucose
▪ Antihyperlipidemic effects Improvement of liver functions
[64,65,66,67]
Summer savorySatureja hortensis L.Aerial parts▪ Stimulation of insulin-dependent glucose uptake▪ Reduction in blood glucose[68]
ThymeThymus vulgaris L.Aerial parts▪ α-glucosidase inhibition
▪ α-amylase inhibition
▪ Stimulation of insulin-dependent glucose uptake
▪ Decrease of fat accumulation in Caenorhabditis elegans
▪ Reduction in blood glucose
▪ Antihyperlipidemic effects
[14,69,70,71]
YarrowAchillea santolina L.Aerial parts▪ α-amylase inhibition▪ Reduction in blood glucose
▪ Antihyperlipidemic effects
[72,73,74]
Table 2. Occurrence of phytochemicals in Mediterranean plant materials and their SMILES notations.
Table 2. Occurrence of phytochemicals in Mediterranean plant materials and their SMILES notations.
CompoundSMILES NotationPlantsReferences
Phenolic acids
Caffeic acidC1=CC(=C(C=C1C=CC(=O)O)O)Oblack mustard, broadleaf plantain, citrus peel, coriander, cumin, dill, garlic, grape skin, marjoram, olive leaf, parsley, rosemary, sage, santolina, thyme[43,75,76,77,78,79,80,81,82,83,84,85]
Cinnamic AcidC1=CC=C(C=C1)C=CC(=O)Odill, grape skin, marjoram, olive leaf, sage, thyme[43,81,82,83,86,87]
Ellagic acidC1=C2C3=C(C(=C1O)O)OC(=O)C4=CC(=C(C(=C43)OC2=O)O)Oblack mustard, broadleaf plantain, citrus fruits, grape skin, parsley, strawberry[75,88,89,90,91,92]
Ferulic acidCOC1=C(C=CC(=C1)C=CC(=O)O)Oblack cumin, citrus peel, coriander, cumin, dill, garlic, marjoram, olive leaf, onion, rosemary, sage, santolina, thyme, strawberry[43,77,78,79,82,85,87,93,94,95,96]
Gallic acidC1=C(C=C(C(=C1O)O)O)C(=O)Oblack mustard, black cumin, citrus peel, cumin, dill, garlic, grape skin and seeds, marjoram, olive leaf, onion, strawberry,[43,76,77,82,86,93,94,97,98,99,100,101]
p-Coumaric acidC1=CC(=CC=C1C=CC(=O)O)Oblack cumin, broadleaf plantain, citrus peel, coriander, cumin, dill, garlic, grape skin, marjoram, olive leaf, parsley, sage, santolina, thyme[43,76,77,78,79,80,81,82,84,85,87,93,99,102]
Rosmarinic acidC1=CC(=C(C=C1CC(C(=O)O)OC(=O)C=CC2=CC(=C(C=C2)O)O)O)Ocumin, dill marjoram, olive leaf, rosemary, sage, santolina, summer savory, thyme[43,82,83,97,102,103,104]
Flavonoids
ApigeninC1=CC(=CC=C1C2=CC(=O)C3=C(C=C(C=C3O2)O)O)Oblack mustard, black cumin, broadleaf plantain, citrus peel, cumin, garlic, marjoram, olive leaf, parsley, santolina, sage, sow thistle, summer savory, thyme,[43,75,76,80,82,85,87,93,97,104,105,106,107]
CatechinC1C(C(OC2=CC(=CC(=C21)O)O)C3=CC(=C(C=C3)O)O)Oblack mustard, black cumin, broadleaf plantain, citrus peel, coriander, cumin, grape skin, olive leaf, onion, strawberry[77,78,82,93,100,108,109,110,111]
ChrysoeriolCOC1=C(C=CC(=C1)C2=CC(=O)C3=C(C=C(C=C3O2)O)O)Obroadleaf plantain, citrus peel, coriander, olive leaf, parsley,[112,113,114,115,116]
HesperidinCC1C(C(C(C(O1)OCC2CC(CC(O2)OC3=CC(=C4C(=O)CC(OC4=C3)C5=CC(=C(C=C5)OΨ)O)O)O)O)O)O)O)Ocitrus peels, marjoram, olive leaf, rosemary, strawberry, summer savory[43,77,82,117,118,119]
KaempferolC1=CC(=CC=C1C2=C(C(=O)C3=C(C=C(C=C3O2)O)O)O)Oblack mustard, citrus peel, coriander, cumin, grape skin, onion, parsley, rosemary, santolina, strawberry, summer savory, thyme[75,77,78,85,94,99,101,120,121,122,123]
LuteolinC1=CC(=C(C=C1C2=CC(=O)C3=C(C=C(C=C3O2)O)O)O)Obroadleaf plantain, citrus peel, coriander, cumin, garlic, marjoram, olive leaf, parsley, rosemary, sage, santolina, sow thistle, strawberry, summer savory, thyme[123,124,125,126,127]
QuercetinC1=CC(=C(C=C1C2=C(C(=O)C3=C(C=C(C=C3O2)O)O)O)O)Oblack mustard, broadleaf plantain, citrus peel, coriander, cumin, dill, garlic, grape skin, marjoram, olive leaf, onion, parsley, rosemary, sage, sow thistle, summer savory, strawberry, thyme[85,92,93,95,96,97,98,102,109,110,117,122,124,125,126,127,128,129]
RutinCC1C(C(C(C(O1)OCC2C(C(C(C(O2)OC3=C(OC4=CC(=CC(=C4C3=O)O)O)C5=CC(=C(C=C5)O)O)O)O)O)O)O)O black mustard, broadleaf plantain, citrus peel, dill, garlic, marjoram, olive leaf, rosemary, sage, santolina, strawberry, summer savory,[90,91,92,97,98,101,102,111,114,122,129,130]
Terpenes
alpha-pineneCC1=CCC2CC1C2(C)Cblack mustard, citrus peel, coriander, cumin, dill, grape skins, marjoram, olive leaf, rosemary, sage, santolina, summer savory, thyme[43,105,131,132,133,134,135,136,137,138,139]
alpha-terpineneCC1=CC=C(CC1)C(C)Ccumin, marjoram, parsley, rosemary, sage, santolina, summer savory, thyme[43,140,141,142,143,144,145,146]
beta-pineneCC1(C2CCC(=C)C1C2)Ccitrus peel, coriander, cumin, grape skins, marjoram, olive leaf, rosemary, sage, summer savory, thyme[43,77,133,134,136,137,138,139]
campheneCC1(C2CCC(C2)C1=C)Ccoriander, marjoram, rosemary, sage, santolina, summer savory, thyme,[43,74,133,138,139]
gamma-terpineneCC1=CCC(=CC1)C(C)Ccitrus peel, coriander, dill, marjoram, parsley, rosemary, sage, santolina, summer savory, thyme[43,77,135,139,141,142,143,144,145,146,147]
limoneneCC1=CCC(CC1)C(=C)Cblack mustard, citrus peel, coriander, dill, marjoram, onion, rosemary, santolina, summer savory, thyme[43,77,132,133,135,139,142,145,146]
beta-myrceneCC(=CCCC(=C)C=C)Ccitrus peel, coriander, dill, grape skins, marjoram, olive leaf, parsley, rosemary, sage, summer savory, thyme[43,105,133,135,136,137,138,139,141,145]
sabineneCC(C)C12CCC(=C)C1C2citrus peel, coriander, marjoram, olive leaf, parsley, sage, santolina, summer savory, thyme[43,74,105,133,137,139,141,143,145]
Table 3. The docking cut-off score of active phytochemicals for each protein target. The score is given in parentheses and expressed as kcal mol−1.
Table 3. The docking cut-off score of active phytochemicals for each protein target. The score is given in parentheses and expressed as kcal mol−1.
Protein TargetPDB CodeFunctionPhytochemicals
Regulation of insulin secretion and/or sensitivity
DPP44A5SStimulation of insulin secretion from pancreas degrading and inactivating glucagon-like peptide-1 [148]Apigenin (−8.2), catechin (−8.3), chrysoeriol (−8.1), ellagic acid (−8.3), hesperidin (−10.4), kaempferol (−8.4), quercetin (−8.3), rutin (−9.1)
FFAR14PHUBinding of free fatty acids to receptor results in increase in glucose-stimulated insulin secretion [149]Apigenin (−8.4), caffeic acid (−8.0), ferulic acid (−8.0), hesperidin (−8.7), luteolin (−8.2)
HSD11B14K1LActivates the synthesis of active glucocorticoids [150]Apigenin (−9.0), catechin (−9.0), chrysoeriol (−9.1), ellagic acid (−8.6), hesperidin (−9.9), kaempferol (−9.2), luteolin (−9.4), quercetin (−9.8), rutin (−9.7)
INSR3EKNRegulates glucose uptake and synthesis of glycogen, lipid, and protein [151]Ellagic acid (−8.2), hesperidin (−9.3), rutin (−8.4)
PTPN94GE6Reduces insulin sensitivity, dephosphorylating the insulin receptor [152]Ellagic acid (−8.0), hesperidin (−8.8), rutin (−8.6)
RBP42WR6Reduces insulin signaling and promotes gluconeogenesis [153]Apigenin (−9.9), catechin (−9.0), chrysoeriol (−9.6), ellagic acid (−8.7), kaempferol (−9.5), luteolin (−9.9), quercetin (−9.6)
Regulation of glucose metabolism
AKR1B13G5ECatalyzes the reduction of glucose to sorbitol [154]Apigenin (−9.1), catechin (−9.1), chrysoeriol (−9.0), ellagic acid (−8.8), hesperidin (−8.1), kaempferol (−8.6), luteolin (−9.1), quercetin (−8.8)
AMY2A4GQRRegulates the digestion of starch to glucose [155]Apigenin (−8.3), catechin (−8.4), chrysoeriol (−8.5), hesperidin (−8.9), kaempferol (−8.0), luteolin (−9.4), quercetin (−9.8), rutin (−9.0)
GCK3IMXPhosphorylates glucose for glycolysis or synthesis of glycogen [156]Hesperidin (−10.2), rutin (−8.6)
MGAM3L4YRegulates the digestion of starch to glucose [157]Hesperidin (−8.2), rutin (−8.3)
PDK24MPCRegulates glucose oxidation through the inactivation of pyruvate dehydrogenase complex [158]Hesperidin (−9.1), rutin (−8.1)
PYGL3DDSRegulates phosphorolysis of glycogen in glycogenesis [159]Hesperidin (−8.6), rutin (−8.6)
Regulation of lipid metabolism
NR5A24DORRegulates the expression of genes involved in the synthesis of bile acid and cholesterol, and steroidogenesis [160]Hesperidin (−8.4)
PPARA3FEIRegulates the expression of genes involved in lipid metabolism [161]Rutin (−8.4)
PPARD3PEQRegulates the expression of genes involved in fatty acid catabolism [162]Chrysoeriol (−8.0), hesperidin (−9.1), rutin (−8.9)
PPARG2FVJRegulates the expression of genes involved in adipogenesis and lipid oxidation [163]Apigenin (−8.1), catechin (−8.2), chrysoeriol (−8.4), ellagic acid (−8.3), hesperidin (−1035), kaempferol (−8.5), luteolin (−8.2), quercetin (−8.4), rutin (−9.8)
RXPRA1FM9Heterodimerizes with PPARs [164]Apigenin (−9.2), chrysoeriol (−9.3), hesperidin (−8.0), luteolin (−9.1)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Goulas, V.; Banegas-Luna, A.J.; Constantinou, A.; Pérez-Sánchez, H.; Barbouti, A. Computation Screening of Multi-Target Antidiabetic Properties of Phytochemicals in Common Edible Mediterranean Plants. Plants 2022, 11, 1637. https://0-doi-org.brum.beds.ac.uk/10.3390/plants11131637

AMA Style

Goulas V, Banegas-Luna AJ, Constantinou A, Pérez-Sánchez H, Barbouti A. Computation Screening of Multi-Target Antidiabetic Properties of Phytochemicals in Common Edible Mediterranean Plants. Plants. 2022; 11(13):1637. https://0-doi-org.brum.beds.ac.uk/10.3390/plants11131637

Chicago/Turabian Style

Goulas, Vlasios, Antonio J. Banegas-Luna, Athena Constantinou, Horacio Pérez-Sánchez, and Alexandra Barbouti. 2022. "Computation Screening of Multi-Target Antidiabetic Properties of Phytochemicals in Common Edible Mediterranean Plants" Plants 11, no. 13: 1637. https://0-doi-org.brum.beds.ac.uk/10.3390/plants11131637

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop